1
|
Migliorini A, Ge S, Atkins MH, Oakie A, Sambathkumar R, Kent G, Huang H, Sing A, Chua C, Gehring AJ, Keller GM, Notta F, Nostro MC. Embryonic macrophages support endocrine commitment during human pancreatic differentiation. Cell Stem Cell 2024; 31:1591-1611.e8. [PMID: 39406230 DOI: 10.1016/j.stem.2024.09.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/02/2024] [Accepted: 09/12/2024] [Indexed: 11/10/2024]
Abstract
Organogenesis is a complex process that relies on a dynamic interplay between extrinsic factors originating from the microenvironment and tissue-specific intrinsic factors. For pancreatic endocrine cells, the local niche consists of acinar and ductal cells as well as neuronal, immune, endothelial, and stromal cells. Hematopoietic cells have been detected in human pancreas as early as 6 post-conception weeks, but whether they play a role during human endocrinogenesis remains unknown. To investigate this, we performed single-nucleus RNA sequencing (snRNA-seq) of the second-trimester human pancreas and identified a wide range of hematopoietic cells, including two distinct subsets of tissue-resident macrophages. Leveraging this discovery, we developed a co-culture system of human embryonic stem cell-derived endocrine-macrophage organoids to model their interaction in vitro. Here, we show that macrophages support the differentiation and viability of endocrine cells in vitro and enhance tissue engraftment, highlighting their potential role in tissue engineering strategies for diabetes.
Collapse
Affiliation(s)
- Adriana Migliorini
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada.
| | - Sabrina Ge
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Michael H Atkins
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Amanda Oakie
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | | | - Gregory Kent
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Haiyang Huang
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Angel Sing
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Conan Chua
- Toronto Centre for Liver Disease, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2C4, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Adam J Gehring
- Toronto Centre for Liver Disease, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2C4, Canada; Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Gordon M Keller
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Faiyaz Notta
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| | - Maria Cristina Nostro
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2C4, Canada.
| |
Collapse
|
2
|
Varghese SS, Hernandez-De La Peña AG, Dhawan S. Safeguarding genomic integrity in beta-cells: implications for beta-cell differentiation, growth, and dysfunction. Biochem Soc Trans 2024; 52:2133-2144. [PMID: 39364746 DOI: 10.1042/bst20231519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
The maintenance of optimal glucose levels in the body requires a healthy reserve of the insulin producing pancreatic beta-cells. Depletion of this reserve due to beta-cell dysfunction and death results in development of diabetes. Recent findings highlight unresolved DNA damage as a key contributor to beta-cell defects in diabetes. Beta-cells face various stressors and metabolic challenges throughout life, rendering them susceptible to DNA breaks. The post-mitotic, long-lived phenotype of mature beta-cells further warrants robust maintenance of genomic integrity. Failure to resolve DNA damage during beta-cell development, therefore, can result in an unhealthy reserve of beta-cells and predispose to diabetes. Yet, the molecular mechanisms safeguarding beta-cell genomic integrity remain poorly understood. Here, we focus on the significance of DNA damage in beta-cell homeostasis and postulate how cellular expansion, epigenetic programming, and metabolic shifts during development may impact beta-cell genomic integrity and health. We discuss recent findings demonstrating a physiological role for DNA breaks in modulating transcriptional control in neurons, which share many developmental programs with beta-cells. Finally, we highlight key gaps in our understanding of beta-cell genomic integrity and discuss emerging areas of interest.
Collapse
Affiliation(s)
- Sneha S Varghese
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA 91010, U.S.A
| | | | - Sangeeta Dhawan
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA 91010, U.S.A
| |
Collapse
|
3
|
Cilvik SN, Boehmer B, Wesolowski SR, Brown LD, Rozance PJ. Chronic late gestation fetal hyperglucagonaemia results in lower insulin secretion, pancreatic mass, islet area and beta- and α-cell proliferation. J Physiol 2024. [PMID: 39383208 DOI: 10.1113/jp286974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 09/09/2024] [Indexed: 10/11/2024] Open
Abstract
Fetal glucagon concentrations are elevated in the presence of a compromised intrauterine environment, as in cases of placental insufficiency and perinatal acidaemia. Our objective was to investigate the impact of late gestation fetal hyperglucagonaemia on in vivo insulin secretion and pancreatic islet structure. Chronically catheterized late gestation fetal sheep received an intravenous infusion of glucagon at low (5 ng/kg/min; GCG-5) or high (50 ng/kg/min; GCG-50) concentrations or a vehicle control (CON) for 8-10 days. Glucose-stimulated fetal insulin secretion (GSIS) was measured following 3 h (acute response) and 8-10 days (chronic response) of experimental infusions. Insulin, glucose and amino acid concentrations were measured longitudinally. The pancreas was collected at the study end for histology and gene expression analysis. Acute exposure (3 h) to GCG-50 induced a 3-fold increase in basal insulin concentrations with greater GSIS. Meanwhile, chronic exposure to both GCG-5 and GCG-50 decreased basal insulin concentrations 2-fold by day 8-10. Chronic GCG-50 also blunted GSIS at the study end. Fetal amino acid concentrations were decreased within 24 h of GCG-5 and GCG-50, while there were no differences in fetal glucose. Histologically, GCG-5 and GCG-50 had lower β- and α-cell proliferation, as well as lower α-cell mass and pancreas weight, while GCG-50 had lower islet area. This study demonstrates that chronic glucagon elevation in late gestation fetuses impairs β-cell proliferation and insulin secretion, which has the potential to contribute to later-life diabetes risk. We speculate that the action of glucagon in lower circulating fetal amino acid concentrations may have a suppressive effect on insulin secretion. KEY POINTS: We have previously demonstrated in a chronically catheterized fetal sheep model that experimentally elevated glucagon in the fetus impairs placental function, reduces fetal protein accretion and lowers fetal weight. In the present study, we further characterized the effects of elevated fetal glucagon on fetal physiology with a focus on pancreatic development and β-cell function. We show that experimentally elevated fetal glucagon results in lower β- and α-cell proliferation, as well as decreased insulin secretion after 8-10 days of glucagon infusion. These results have important implications for β-cell reserve and later-life predisposition to diabetes.
Collapse
Affiliation(s)
- Sarah N Cilvik
- Perinatal Research Center, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Brit Boehmer
- Perinatal Research Center, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Stephanie R Wesolowski
- Perinatal Research Center, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Laura D Brown
- Perinatal Research Center, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Paul J Rozance
- Perinatal Research Center, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
4
|
Barco VS, Gallego FQ, Miranda CA, Souza MR, Volpato GT, Damasceno DC. Hyperglycemia influences the cell proliferation and death of the rat endocrine pancreas in the neonatal period. Life Sci 2024; 351:122854. [PMID: 38901688 DOI: 10.1016/j.lfs.2024.122854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/07/2024] [Accepted: 06/16/2024] [Indexed: 06/22/2024]
Abstract
AIMS To evaluate the cell proliferation and death, and structural morphology of the pancreatic islet cells of the rats with hyperglycemia in the first month of life and compare to those of the control rats. MAIN METHODS Female Sprague-Dawley newborn rats received Streptozotocin (a beta-cytotoxic drug) at birth for diabetes induction. Control and hyperglycemic animals were euthanized on different days of life: 5, 10, 15, and 30. The pancreas was collected and processed for immunohistochemical analysis of cleaved Caspase-3 (cell death), Ki-67 (cell proliferation), PDX-1 (transcription factor responsible for insulin synthesis), and endocrine hormones (insulin, glucagon, and somatostatin). KEY FINDINGS Control females showed a higher percentage (%) of Ki-67-positive(+) cells on D10 and D15, a higher % of insulin+ and somatostatin+ cells on D15 and D30, a lower % of PDX-1+ cells on D10, and a higher % of glucagon+ cells on D10 and D30. Hyperglycemic females showed a lower % of Ki-67+ cells on D15, a higher % of cleaved Caspase-3+ cells on D15, and insulin+ cells on D15 and D30. In the comparison among the experimental groups, the hyperglycemic females showed an increased % of cleaved Caspase-3+ and Ki-67+ cells and a lower % of PDX-1+ cells. SIGNIFICANCE This study enabled a better understanding of the abnormal pancreas development regarding cellular proliferation, apoptosis, and hormonal synthesis in the neonatal period. Thus, the pancreatic islets of hyperglycemic rats do not reestablish the normal endocrine cell population, and cellular apoptosis overcame the proliferative activity of these cells.
Collapse
Affiliation(s)
- Vinícius S Barco
- Laboratory of Experimental Research on Gynecology and Obstetrics of UNIPEX, Postgraduate Course on Tocogynecology, Botucatu Medical School, Sao Paulo State University (Unesp), Botucatu, Sao Paulo State, Brazil.
| | - Franciane Q Gallego
- Laboratory of Experimental Research on Gynecology and Obstetrics of UNIPEX, Postgraduate Course on Tocogynecology, Botucatu Medical School, Sao Paulo State University (Unesp), Botucatu, Sao Paulo State, Brazil.
| | - Carolina A Miranda
- Laboratory of Experimental Research on Gynecology and Obstetrics of UNIPEX, Postgraduate Course on Tocogynecology, Botucatu Medical School, Sao Paulo State University (Unesp), Botucatu, Sao Paulo State, Brazil
| | - Maysa R Souza
- Laboratory of Experimental Research on Gynecology and Obstetrics of UNIPEX, Postgraduate Course on Tocogynecology, Botucatu Medical School, Sao Paulo State University (Unesp), Botucatu, Sao Paulo State, Brazil.
| | - Gustavo T Volpato
- Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso State, Brazil
| | - Débora C Damasceno
- Laboratory of Experimental Research on Gynecology and Obstetrics of UNIPEX, Postgraduate Course on Tocogynecology, Botucatu Medical School, Sao Paulo State University (Unesp), Botucatu, Sao Paulo State, Brazil.
| |
Collapse
|
5
|
Kowalska M, Kaczmarek P, Rupik W. Does the pancreas of gekkotans differentiate similarly? Developmental structural and 3D studies of the mourning gecko (Lepidodactylus lugubris) and the leopard gecko (Eublepharis macularius). J Anat 2024; 245:303-323. [PMID: 38581181 PMCID: PMC11259754 DOI: 10.1111/joa.14038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/12/2024] [Accepted: 03/10/2024] [Indexed: 04/08/2024] Open
Abstract
This study investigated the pancreas differentiation of two species of gekkotan families-the mourning gecko Lepidodactylus lugubris (Gekkonidae) and the leopard gecko Eublepharis macularius (Eublepharidae)-based on two-dimensional (2D) histological samples and three-dimensional (3D) reconstructions of the position of the pancreatic buds and the surrounding organs. The results showed that at the moment of egg laying, the pancreas of L. lugubris is composed of three distinct primordia: one dorsal and two ventral. The dorsal primordium differentiates earlier than either ventral primordium. The right ventral primordium is more prominent and distinctive, starting to form earlier than the left one. Moreover, at this time, the pancreas of the leopard gecko is composed of the dorsal and right ventral primordium and the duct of the left ventral primordium. It means that the leopard gecko's left primordium is a transitional structure. These results indicate that the early development of the gekkotan pancreas is species specific. The pancreatic buds of the leopard and mourning gecko initially enter the duodenum by separate outlets, similar to the pancreas of other vertebrates. The pancreatic buds (3 of the mourning gecko and 2 of the leopard gecko) fuse quickly and form an embryonic pancreas. After that, the structure of this organ changes. After fusion, the pancreas of both gekkotans comprises four parts: the head of the pancreas (central region) and three lobes: upper, splenic, and lower. This organ develops gradually and is very well distinguished at hatching time. In both gekkotan species, cystic, hepatic, and pancreatic ducts enter the duodenum within the papilla. During gekkotan pancreas differentiation, the connection between the common bile duct and the dorsal pancreatic duct is associated with intestinal rotation, similar to other vertebrates.
Collapse
Affiliation(s)
- Magdalena Kowalska
- Institute of Biology, Biotechnology and Environmental Protection, Faculty of Natural Sciences, University of Silesia in Katowice, Katowice, Poland
| | - Paweł Kaczmarek
- Institute of Biology, Biotechnology and Environmental Protection, Faculty of Natural Sciences, University of Silesia in Katowice, Katowice, Poland
| | - Weronika Rupik
- Institute of Biology, Biotechnology and Environmental Protection, Faculty of Natural Sciences, University of Silesia in Katowice, Katowice, Poland
| |
Collapse
|
6
|
Zhu J, Zhu X, Xu Y, Chen X, Ge X, Huang Y, Wang Z. The role of noncoding RNAs in beta cell biology and tissue engineering. Life Sci 2024; 348:122717. [PMID: 38744419 DOI: 10.1016/j.lfs.2024.122717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/29/2024] [Accepted: 05/11/2024] [Indexed: 05/16/2024]
Abstract
The loss or dysfunction of pancreatic β-cells, which are responsible for insulin secretion, constitutes the foundation of all forms of diabetes, a widely prevalent disease worldwide. The replacement of damaged β-cells with regenerated or transplanted cells derived from stem cells is a promising therapeutic strategy. However, inducing the differentiation of stem cells into fully functional glucose-responsive β-cells in vitro has proven to be challenging. Noncoding RNAs (ncRNAs) have emerged as critical regulatory factors governing the differentiation, identity, and function of β-cells. Furthermore, engineered hydrogel systems, biomaterials, and organ-like structures possess engineering characteristics that can provide a three-dimensional (3D) microenvironment that supports stem cell differentiation. This review summarizes the roles and contributions of ncRNAs in maintaining the differentiation, identity, and function of β-cells. And it focuses on regulating the levels of ncRNAs in stem cells to activate β-cell genetic programs for generating alternative β-cells and discusses how to manipulate ncRNA expression by combining hydrogel systems and other tissue engineering materials. Elucidating the patterns of ncRNA-mediated regulation in β-cell biology and utilizing this knowledge to control stem cell differentiation may offer promising therapeutic strategies for generating functional insulin-producing cells in diabetes cell replacement therapy and tissue engineering.
Collapse
Affiliation(s)
- Jiaqi Zhu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Xiaoren Zhu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Yang Xu
- Center of Gallbladder Disease, Shanghai East Hospital, Institute of Gallstone Disease, School of Medicine, Tongji University, Shanghai 200092, China
| | - Xingyou Chen
- Medical School of Nantong University, Nantong 226001, China
| | - Xinqi Ge
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Yan Huang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| | - Zhiwei Wang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| |
Collapse
|
7
|
Villalba A, Gitton Y, Inoue M, Aiello V, Blain R, Toupin M, Mazaud-Guittot S, Rachdi L, Semb H, Chédotal A, Scharfmann R. A 3D atlas of the human developing pancreas to explore progenitor proliferation and differentiation. Diabetologia 2024; 67:1066-1078. [PMID: 38630142 PMCID: PMC11058870 DOI: 10.1007/s00125-024-06143-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/07/2024] [Indexed: 04/30/2024]
Abstract
AIMS/HYPOTHESIS Rodent pancreas development has been described in great detail. On the other hand, there are still gaps in our understanding of the developmental trajectories of pancreatic cells during human ontogenesis. Here, our aim was to map the spatial and chronological dynamics of human pancreatic cell differentiation and proliferation by using 3D imaging of cleared human embryonic and fetal pancreases. METHODS We combined tissue clearing with light-sheet fluorescence imaging in human embryonic and fetal pancreases during the first trimester of pregnancy. In addition, we validated an explant culture system enabling in vitro proliferation of pancreatic progenitors to determine the mitogenic effect of candidate molecules. RESULTS We detected the first insulin-positive cells as early as five post-conceptional weeks, two weeks earlier than previously observed. We observed few insulin-positive clusters at five post-conceptional weeks (mean ± SD 9.25±5.65) with a sharp increase to 11 post-conceptional weeks (4307±152.34). We identified a central niche as the location of onset of the earliest insulin cell production and detected extra-pancreatic loci within the adjacent developing gut. Conversely, proliferating pancreatic progenitors were located in the periphery of the epithelium, suggesting the existence of two separated pancreatic niches for differentiation and proliferation. Additionally, we observed that the proliferation ratio of progenitors ranged between 20% and 30%, while for insulin-positive cells it was 1%. We next unveiled a mitogenic effect of the platelet-derived growth factor AA isoform (PDGFAA) in progenitors acting through the pancreatic mesenchyme by increasing threefold the number of proliferating progenitors. CONCLUSIONS/INTERPRETATION This work presents a first 3D atlas of the human developing pancreas, charting both endocrine and proliferating cells across early development.
Collapse
Affiliation(s)
- Adrian Villalba
- Institut Cochin, CNRS, Inserm, Université Paris Cité, Paris, France
| | - Yorick Gitton
- Inserm, CNRS, Institut de la Vision, Sorbonne Université, Paris, France
| | - Megumi Inoue
- Inserm, CNRS, Institut de la Vision, Sorbonne Université, Paris, France
| | - Virginie Aiello
- Institut Cochin, CNRS, Inserm, Université Paris Cité, Paris, France
| | - Raphaël Blain
- Inserm, CNRS, Institut de la Vision, Sorbonne Université, Paris, France
| | - Maryne Toupin
- Inserm, EHESP, IRSET (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, Université Rennes, Rennes, France
| | - Séverine Mazaud-Guittot
- Inserm, EHESP, IRSET (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, Université Rennes, Rennes, France
| | - Latif Rachdi
- Institut Cochin, CNRS, Inserm, Université Paris Cité, Paris, France
| | - Henrik Semb
- Institute of Translational Stem Cell Research, Helmholtz Diabetes Center, Helmholtz Zentrum München, München, Germany
| | - Alain Chédotal
- Inserm, CNRS, Institut de la Vision, Sorbonne Université, Paris, France.
- Institut de pathologie, groupe hospitalier Est, hospices civils de Lyon, Lyon, France.
- MeLiS, CNRS UMR5284, Inserm U1314, University Claude Bernard Lyon 1, Lyon, France.
| | | |
Collapse
|
8
|
Saloman JL, Epouhe AY, Ruff CF, Albers KM. PDX1, a transcription factor essential for organ differentiation, regulates SERCA-dependent Ca 2+ homeostasis in sensory neurons. Cell Calcium 2024; 120:102884. [PMID: 38574509 PMCID: PMC11188734 DOI: 10.1016/j.ceca.2024.102884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/13/2024] [Accepted: 03/31/2024] [Indexed: 04/06/2024]
Abstract
Pancreatic and duodenal homeobox 1 (PDX1) is a transcription factor required for the development and differentiation of the pancreas. Previous studies indicated that PDX1 expression was restricted to the gastrointestinal tract. Using a cre-dependent reporter, we observed PDX1-dependent expression of tdtomato (PDX1-tom) in a subpopulation of sensory nerves. Many of these PDX1-tom afferents expressed the neurofilament 200 protein and projected to the skin. Tdtomato-labeled terminals were associated with hair follicles in the form of longitudinal and circumferential lanceolate endings suggesting a role in tactile and proprioceptive perception. To begin to examine the functional significance of PDX1 in afferents, we used Fura-2 imaging to examine calcium (Ca2+) handling under naïve and nerve injury conditions. Neuropathic injury is associated with increased intracellular Ca2+ signaling that in part results from dysregulation of the sarco/endoplasmic reticulum calcium transport ATPase (SERCA). Here we demonstrate that under naïve conditions, PDX1 regulates expression of the SERCA2B isoform in sensory neurons. In response to infraorbital nerve injury, a significant reduction of PDX1 and SERCA2B expression and dysregulation of Ca2+ handling occurs in PDX1-tom trigeminal ganglia neurons. The identification of PDX1 expression in the somatosensory system and its regulation of SERCA2B and Ca2+ handling provide a new mechanism to explain pathological changes in primary afferents that may contribute to pain associated with nerve injury.
Collapse
Affiliation(s)
- Jami L Saloman
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Neurobiology, Center for Neuroscience and Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Ariel Y Epouhe
- Department of Neurobiology, Center for Neuroscience and Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Catherine F Ruff
- Department of Neurobiology, Center for Neuroscience and Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kathryn M Albers
- Department of Neurobiology, Center for Neuroscience and Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
9
|
Kennedy VC, Lynch CS, Tanner AR, Winger QA, Gad A, Rozance PJ, Anthony RV. Fetal Hypoglycemia Induced by Placental SLC2A3-RNA Interference Alters Fetal Pancreas Development and Transcriptome at Mid-Gestation. Int J Mol Sci 2024; 25:4780. [PMID: 38731997 PMCID: PMC11084495 DOI: 10.3390/ijms25094780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Glucose, the primary energy substrate for fetal oxidative processes and growth, is transferred from maternal to fetal circulation down a concentration gradient by placental facilitative glucose transporters. In sheep, SLC2A1 and SLC2A3 are the primary transporters available in the placental epithelium, with SLC2A3 located on the maternal-facing apical trophoblast membrane and SLC2A1 located on the fetal-facing basolateral trophoblast membrane. We have previously reported that impaired placental SLC2A3 glucose transport resulted in smaller, hypoglycemic fetuses with reduced umbilical artery insulin and glucagon concentrations, in addition to diminished pancreas weights. These findings led us to subject RNA derived from SLC2A3-RNAi (RNA interference) and NTS-RNAi (non-targeting sequence) fetal pancreases to qPCR followed by transcriptomic analysis. We identified a total of 771 differentially expressed genes (DEGs). Upregulated pathways were associated with fat digestion and absorption, particularly fatty acid transport, lipid metabolism, and cholesterol biosynthesis, suggesting a potential switch in energetic substrates due to hypoglycemia. Pathways related to molecular transport and cell signaling in addition to pathways influencing growth and metabolism of the developing pancreas were also impacted. A few genes directly related to gluconeogenesis were also differentially expressed. Our results suggest that fetal hypoglycemia during the first half of gestation impacts fetal pancreas development and function that is not limited to β cell activity.
Collapse
Affiliation(s)
- Victoria C. Kennedy
- College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA; (V.C.K.); (C.S.L.); (A.R.T.); (Q.A.W.); (A.G.)
| | - Cameron S. Lynch
- College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA; (V.C.K.); (C.S.L.); (A.R.T.); (Q.A.W.); (A.G.)
| | - Amelia R. Tanner
- College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA; (V.C.K.); (C.S.L.); (A.R.T.); (Q.A.W.); (A.G.)
- University of Colorado School of Medicine, Aurora, CO 80045, USA;
| | - Quinton A. Winger
- College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA; (V.C.K.); (C.S.L.); (A.R.T.); (Q.A.W.); (A.G.)
| | - Ahmed Gad
- College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA; (V.C.K.); (C.S.L.); (A.R.T.); (Q.A.W.); (A.G.)
| | - Paul J. Rozance
- University of Colorado School of Medicine, Aurora, CO 80045, USA;
| | - Russell V. Anthony
- College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA; (V.C.K.); (C.S.L.); (A.R.T.); (Q.A.W.); (A.G.)
| |
Collapse
|
10
|
Oropeza D, Herrera PL. Glucagon-producing α-cell transcriptional identity and reprogramming towards insulin production. Trends Cell Biol 2024; 34:180-197. [PMID: 37626005 DOI: 10.1016/j.tcb.2023.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 08/27/2023]
Abstract
β-Cell replacement by in situ reprogramming of non-β-cells is a promising diabetes therapy. Following the observation that near-total β-cell ablation in adult mice triggers the reprogramming of pancreatic α-, δ-, and γ-cells into insulin (INS)-producing cells, recent studies are delving deep into the mechanisms controlling adult α-cell identity. Systematic analyses of the α-cell transcriptome and epigenome have started to pinpoint features that could be crucial for maintaining α-cell identity. Using different transgenic and chemical approaches, significant advances have been made in reprogramming α-cells in vivo into INS-secreting cells in mice. The recent reprogramming of human α-cells in vitro is an important step forward that must now be complemented with a comprehensive molecular dissection of the mechanisms controlling α-cell identity.
Collapse
Affiliation(s)
- Daniel Oropeza
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Pedro Luis Herrera
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
11
|
Uddin MH, Zhang D, Muqbil I, El-Rayes BF, Chen H, Philip PA, Azmi AS. Deciphering cellular plasticity in pancreatic cancer for effective treatments. Cancer Metastasis Rev 2024; 43:393-408. [PMID: 38194153 DOI: 10.1007/s10555-023-10164-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 12/19/2023] [Indexed: 01/10/2024]
Abstract
Cellular plasticity and therapy resistance are critical features of pancreatic cancer, a highly aggressive and fatal disease. The pancreas, a vital organ that produces digestive enzymes and hormones, is often affected by two main types of cancer: the pre-dominant ductal adenocarcinoma and the less common neuroendocrine tumors. These cancers are difficult to treat due to their complex biology characterized by cellular plasticity leading to therapy resistance. Cellular plasticity refers to the capability of cancer cells to change and adapt to different microenvironments within the body which includes acinar-ductal metaplasia, epithelial to mesenchymal/epigenetic/metabolic plasticity, as well as stemness. This plasticity allows heterogeneity of cancer cells, metastasis, and evasion of host's immune system and develops resistance to radiation, chemotherapy, and targeted therapy. To overcome this resistance, extensive research is ongoing exploring the intrinsic and extrinsic factors through cellular reprogramming, chemosensitization, targeting metabolic, key survival pathways, etc. In this review, we discussed the mechanisms of cellular plasticity involving cellular adaptation and tumor microenvironment and provided a comprehensive understanding of its role in therapy resistance and ways to overcome it.
Collapse
Affiliation(s)
- Md Hafiz Uddin
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, 4100 John R, HWCRC 740, Detroit, MI, 48201, USA.
| | - Dingqiang Zhang
- Department of Natural Sciences, Lawrence Technological University, 21000 W 10 Mile Rd, Southfield, MI, 48075, USA
| | - Irfana Muqbil
- Department of Natural Sciences, Lawrence Technological University, 21000 W 10 Mile Rd, Southfield, MI, 48075, USA
| | - Bassel F El-Rayes
- Division of Hematology and Oncology, Department of Medicine, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, 35233, USA
| | - Herbert Chen
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Philip A Philip
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, 4100 John R, HWCRC 740, Detroit, MI, 48201, USA
- Henry Ford Health Systems, Detroit, MI, 48202, USA
| | - Asfar S Azmi
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, 4100 John R, HWCRC 740, Detroit, MI, 48201, USA.
| |
Collapse
|
12
|
Wu M, Mi J, Qu GX, Zhang S, Jian Y, Gao C, Cai Q, Liu J, Jiang J, Huang H. Role of Hedgehog Signaling Pathways in Multipotent Mesenchymal Stem Cells Differentiation. Cell Transplant 2024; 33:9636897241244943. [PMID: 38695366 PMCID: PMC11067683 DOI: 10.1177/09636897241244943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 03/09/2024] [Accepted: 03/14/2024] [Indexed: 05/05/2024] Open
Abstract
Multipotent mesenchymal stem cells (MSCs) have high self-renewal and multi-lineage differentiation potentials and low immunogenicity, so they have attracted much attention in the field of regenerative medicine and have a promising clinical application. MSCs originate from the mesoderm and can differentiate not only into osteoblasts, cartilage, adipocytes, and muscle cells but also into ectodermal and endodermal cell lineages across embryonic layers. To design cell therapy for replacement of damaged tissues, it is essential to understand the signaling pathways, which have a major impact on MSC differentiation, as this will help to integrate the signaling inputs to initiate a specific lineage. Hedgehog (Hh) signaling plays a vital role in the development of various tissues and organs in the embryo. As a morphogen, Hh not only regulates the survival and proliferation of tissue progenitor and stem populations but also is a critical moderator of MSC differentiation, involving tri-lineage and across embryonic layer differentiation of MSCs. This review summarizes the role of Hh signaling pathway in the differentiation of MSCs to mesodermal, endodermal, and ectodermal cells.
Collapse
Affiliation(s)
- Mengyu Wu
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, China
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Junwei Mi
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, China
| | - Guo-xin Qu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Shu Zhang
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, China
| | - Yi Jian
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, China
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Chu Gao
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, China
| | - Qingli Cai
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, China
| | - Jing Liu
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, China
| | - Jianxin Jiang
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, China
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Hong Huang
- Department of Trauma Medical Center, Daping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, China
| |
Collapse
|
13
|
Sun HY, Lin XY. Genetic perspectives on childhood monogenic diabetes: Diagnosis, management, and future directions. World J Diabetes 2023; 14:1738-1753. [PMID: 38222792 PMCID: PMC10784795 DOI: 10.4239/wjd.v14.i12.1738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 10/10/2023] [Accepted: 11/14/2023] [Indexed: 12/14/2023] Open
Abstract
Monogenic diabetes is caused by one or even more genetic variations, which may be uncommon yet have a significant influence and cause diabetes at an early age. Monogenic diabetes affects 1 to 5% of children, and early detection and gene-tically focused treatment of neonatal diabetes and maturity-onset diabetes of the young can significantly improve long-term health and well-being. The etiology of monogenic diabetes in childhood is primarily attributed to genetic variations affecting the regulatory genes responsible for beta-cell activity. In rare instances, mutations leading to severe insulin resistance can also result in the development of diabetes. Individuals diagnosed with specific types of monogenic diabetes, which are commonly found, can transition from insulin therapy to sulfonylureas, provided they maintain consistent regulation of their blood glucose levels. Scientists have successfully devised materials and methodologies to distinguish individuals with type 1 or 2 diabetes from those more prone to monogenic diabetes. Genetic screening with appropriate findings and interpretations is essential to establish a prognosis and to guide the choice of therapies and management of these interrelated ailments. This review aims to design a comprehensive literature summarizing genetic insights into monogenetic diabetes in children and adolescents as well as summarizing their diagnosis and mana-gement.
Collapse
Affiliation(s)
- Hong-Yan Sun
- Department of Endocrine and Metabolic Diseases, Yantaishan Hospital, Yantai 264003, Shandong Province, China
| | - Xiao-Yan Lin
- Department of Endocrine and Metabolic Diseases, Yantaishan Hospital, Yantai 264003, Shandong Province, China
| |
Collapse
|
14
|
Roy RV, Means N, Rao G, Asfa S, Madka V, Dey A, Zhang Y, Choudhury M, Fung KM, Dhanasekaran DN, Friedman JE, Crawford HC, Rao CV, Bhattacharya R, Mukherjee P. Pancreatic Ubap2 deletion regulates glucose tolerance, inflammation, and protection from cerulein-induced pancreatitis. Cancer Lett 2023; 578:216455. [PMID: 37865160 PMCID: PMC10897936 DOI: 10.1016/j.canlet.2023.216455] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/10/2023] [Accepted: 10/17/2023] [Indexed: 10/23/2023]
Abstract
Ubiquitin-binding associated protein 2 (UBAP2) is reported to promote macropinocytosis and pancreatic adenocarcinoma (PDAC) growth, however, its role in normal pancreatic function remains unknown. We addressed this knowledge gap by generating UBAP2 knockout (U2KO) mice under a pancreas-specific Cre recombinase (Pdx1-Cre). Pancreatic architecture remained intact in U2KO animals, but they demonstrated slight glucose intolerance compared to controls. Upon cerulein challenge to induce pancreatitis, U2KO animals had reduced levels of several pancreatitis-relevant cytokines, amylase and lipase in the serum, reduced tissue damage, and lessened neutrophil infiltration into the pancreatic tissue. Mechanistically, cerulein-challenged U2KO animals revealed reduced NF-κB activation compared to controls. In vitro promoter binding studies confirmed the reduction of NF-κB binding to its target molecules supporting UBAP2 as a new regulator of inflammation in pancreatitis and may be exploited as a therapeutic target in future to inhibit pancreatitis.
Collapse
Affiliation(s)
- Ram Vinod Roy
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Nicolas Means
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Geeta Rao
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Sima Asfa
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Venkateshwar Madka
- Center for Cancer Prevention and Drug Development, Department of Medicine, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anindya Dey
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Yushan Zhang
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Monalisa Choudhury
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kar-Ming Fung
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Danny N Dhanasekaran
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Cell Biology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Jacob E Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Howard C Crawford
- Department of Surgery, Henry Ford Pancreatic Cancer Center, Henry Ford Health System, Detroit, MI, USA
| | - Chinthalapally V Rao
- Center for Cancer Prevention and Drug Development, Department of Medicine, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Resham Bhattacharya
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Obstetrics and Gynecology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Priyabrata Mukherjee
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
15
|
Ortega-Pineda L, Guilfoyle E, Rincon-Benavides MA, Anaparthi AL, Lemmerman LR, Cuellar-Gaviria TZ, Lawrence W, Buss JL, Deng B, Blackstone BN, Salazar-Puerta A, McComb DW, Powell H, Gallego-Perez D, Higuita-Castro N. Engineered extracellular vesicles from human skin cells induce pro-β-cell conversions in pancreatic ductal cells. ADVANCED NANOBIOMED RESEARCH 2023; 3:2200173. [PMID: 38911285 PMCID: PMC11192446 DOI: 10.1002/anbr.202200173] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/25/2024] Open
Abstract
Direct nuclear reprogramming has the potential to enable the development of β cell replacement therapies for diabetes that do not require the use of progenitor/stem cell populations. However, despite their promise, current approaches to β cell-directed reprogramming rely heavily on the use of viral vectors. Here we explored the use of extracellular vesicles (EVs) derived from human dermal fibroblasts (HDFs) as novel non-viral carriers of endocrine cell-patterning transcription factors, to transfect and transdifferentiate pancreatic ductal epithelial cells (PDCs) into hormone-expressing cells. Electrotransfection of HDFs with expression plasmids for Pdx1, Ngn3, and MafA (PNM) led to the release of EVs loaded with PNM at the gene, mRNA, and protein level. Exposing PDC cultures to PNM-loaded EVs led to successful transfection and increased PNM expression in PDCs, which ultimately resulted in endocrine cell-directed conversions based on the expression of insulin/c-peptide, glucagon, and glucose transporter 2 (Glut2). These findings were further corroborated in vivo in a mouse model following intraductal injection of PNM- vs sham-loaded EVs. Collectively these findings suggest that dermal fibroblast-derived EVs could potentially serve as a powerful platform technology for the development and deployment of non-viral reprogramming-based cell therapies for insulin-dependent diabetes.
Collapse
Affiliation(s)
| | - Elizabeth Guilfoyle
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH
| | | | | | - Luke R. Lemmerman
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH
| | | | - William Lawrence
- Biomedical Science Graduate Program, The Ohio State University, Columbus, OH
| | - Jill L Buss
- Department of Hematology and the Bloomfield Center for Leukemia Outcomes Research, The Ohio State University, Columbus, OH
| | - Binbin Deng
- Center for Electron Microscopy and Analysis (CEMAS), The Ohio State University, Columbus, OH
| | - Britani N. Blackstone
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH
| | - Ana Salazar-Puerta
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH
| | - David W. McComb
- Center for Electron Microscopy and Analysis (CEMAS), The Ohio State University, Columbus, OH
| | - Heather Powell
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH
- Shriners Hospitals-Ohio, Dayton, OH 45404, USA
| | - Daniel Gallego-Perez
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH
- Department of Surgery, -The Ohio State University, Columbus, OH
| | - Natalia Higuita-Castro
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH
- Department of Surgery, -The Ohio State University, Columbus, OH
| |
Collapse
|
16
|
Su Q, Yuan F, Li X, Wang X, Yang K, Shao L, Li W. Wfs1 loss-of-function disrupts the composition of mouse pancreatic endocrine cells from birth and impairs Glut2 localization to cytomembrane in pancreatic β cells. Biochem Biophys Res Commun 2023; 658:80-87. [PMID: 37027908 DOI: 10.1016/j.bbrc.2023.03.074] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023]
Abstract
Wfs1 is an endoplasmic reticulum (ER) membrane located protein highly expressed in pancreatic β cells and brain. Wfs1 deficiency causes adult pancreatic β cells dysfunction following β cells apoptosis. Previous studies mainly focus on the Wfs1 function in adult mouse pancreatic β cells. However, whether Wfs1 loss-of-function impairs mouse pancreatic β cell from its early development is unknown. In our study, Wfs1 deficiency disrupts the composition of mouse pancreatic endocrine cells from early postnatal day 0 (P0) to 8 weeks old, with decreased percentage of β cells and increased percentage of α and δ cells. Meanwhile, Wfs1 loss-of-function leads to reduced intracellular insulin content. Notably, Wfs1 deficiency impairs Glut2 localization and causes the accumulation of Glut2 in mouse pancreatic β cell cytoplasm. In Wfs1-deficient mice, glucose homeostasis is disturbed from early 3 weeks old to 8 weeks old. This work reveals that Wfs1 is significantly required for the composition of pancreatic endocrine cells and is essential for Glut2 localization in mouse pancreatic β cells.
Collapse
Affiliation(s)
- Qiang Su
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Fei Yuan
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xiaobo Li
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xuan Wang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Kaijiang Yang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Li Shao
- Department of VIP Clinic, Shanghai East Hospital, Tongji University School of Medicine, No. 1800 Yuntai Road, Pudong District, Shanghai, 200092, China.
| | - Weida Li
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China; Reg-Verse Therapeutics (Shanghai) Co. Ltd., Shanghai, 200120, China.
| |
Collapse
|
17
|
Abstract
Pancreatic ductal adenocarcinomas are distinguished by their robust desmoplasia, or fibroinflammatory response. Dominated by non-malignant cells, the mutated epithelium must therefore combat, cooperate with or co-opt the surrounding cells and signalling processes in its microenvironment. It is proposed that an invasive pancreatic ductal adenocarcinoma represents the coordinated evolution of malignant and non-malignant cells and mechanisms that subvert and repurpose normal tissue composition, architecture and physiology to foster tumorigenesis. The complex kinetics and stepwise development of pancreatic cancer suggests that it is governed by a discrete set of organizing rules and principles, and repeated attempts to target specific components within the microenvironment reveal self-regulating mechanisms of resistance. The histopathological and genetic progression models of the transforming ductal epithelium must therefore be considered together with a programme of stromal progression to create a comprehensive picture of pancreatic cancer evolution. Understanding the underlying organizational logic of the tumour to anticipate and pre-empt the almost inevitable compensatory mechanisms will be essential to eradicate the disease.
Collapse
Affiliation(s)
- Sunil R Hingorani
- Division of Hematology and Oncology, Department of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
- Pancreatic Cancer Center of Excellence, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
18
|
Autophagy-Related ncRNAs in Pancreatic Cancer. Pharmaceuticals (Basel) 2022; 15:ph15121547. [PMID: 36558998 PMCID: PMC9785627 DOI: 10.3390/ph15121547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/05/2022] [Accepted: 12/10/2022] [Indexed: 12/15/2022] Open
Abstract
Pancreatic cancer (PC) is a malignancy accounting for only 3% of total cancers, but with a low 5-year relative survival rate. Approximately 80% of PC patients are diagnosed at a late stage when the disease has already spread from the primary site. Despite advances in PC treatment, there is an urgently needed for the identification of novel therapeutic strategies for PC, particularly for patients who cannot undergo classical surgery. Autophagy is an evolutionarily conserved process used by cells to adapt to metabolic stress via the degrading or recycling of damaged or unnecessary organelles and cellular components. This process is elevated in PC and, thus, it contributes to the onset, progression, and cancer cell resistance to chemotherapy in pancreatic tumors. Autophagy inhibition has been shown to lead to cancer regression and to increase the sensitivity of pancreatic cells to radiation and chemotherapy. Emerging studies have focused on the roles of non-coding RNAs (ncRNAs), such as miRNAs, long non-coding RNAs, and circular RNAs, in PC development and progression. Furthermore, ncRNAs have been reported as crucial regulators of many biological processes, including autophagy, suggesting that ncRNA-based autophagy targeting methods could be promising novel molecular approaches for specifically reducing autophagic flux, thus improving the management of PC patients. In this review, we briefly summarize the existing studies regarding the role and the regulatory mechanisms of autophagy-related ncRNAs in the context of this cancer.
Collapse
|
19
|
Oyedeji O, Rodgers S, Wrubel A, Shah R, Husain S. Pancreatic lymphoepithelial cyst with concurrent HIV infection: A case report and review of the literature. SAGE Open Med Case Rep 2022; 10:2050313X221131166. [PMID: 36274859 PMCID: PMC9580076 DOI: 10.1177/2050313x221131166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/19/2022] [Indexed: 11/15/2022] Open
Abstract
Pancreatic lymphoepithelial cysts are rare, benign, non-neoplastic unilocular or multilocular cystic lesions. These circumscribed pancreatic lesions are filled with keratinous material grossly and exhibit distinct microscopic features. Pancreatic lymphoepithelial cysts are like the more common lymphoepithelial cysts of the parotid glands, which have been associated with the diffuse infiltrative lymphocytosis syndrome often seen in patients with HIV infection. However, pancreatic lymphoepithelial cysts are rare and their association with HIV infection has not been established. The presence of secondary changes in non-neoplastic cysts such as goblet cell metaplasia that was present in our case is an important feature to be included in the differential diagnosis and not to be interpreted as a mucinous neoplasm, particularly on fine-needle aspiration specimen microscopic evaluation that would impact further management. Here we describe the diagnosis and treatment of lymphoepithelial cysts in a patient who was on highly active antiretroviral therapy for HIV infection and we provide a brief literature review. Defining the clinical characteristics of lymphoepithelial cysts in patients with HIV and determining accurate preoperative diagnostic procedures will be critical for establishing effective surgical and medical approaches to treating these cysts, which differ substantially from other more serious pancreatic cystic lesions.
Collapse
Affiliation(s)
- Oluwayomi Oyedeji
- Department of Pathology and Laboratory Medicine, Henry Ford Hospital, Detroit, MI, USA,Oluwayomi Oyedeji, Department of Pathology and Laboratory Medicine, Henry Ford Hospital, Detroit, MI 48202, USA.
| | - Shannon Rodgers
- Department of Pathology and Laboratory Medicine, Henry Ford Hospital, Detroit, MI, USA
| | - Allen Wrubel
- Department of Diagnostic Radiology, Henry Ford Hospital, Detroit, MI, USA
| | - Rupen Shah
- Division of Surgical Oncology, Department of Surgery, Henry Ford Hospital, Detroit, MI, USA
| | - Sanam Husain
- Department of Pathology and Laboratory Medicine, Henry Ford Hospital, Detroit, MI, USA
| |
Collapse
|
20
|
Expression Profiling of Pdx1, Ngn3, and MafA in the Liver and Pancreas of Recovering Streptozotocin-Induced Diabetic Rats. Genes (Basel) 2022; 13:genes13091625. [PMID: 36140793 PMCID: PMC9498460 DOI: 10.3390/genes13091625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
Studies in animal diabetic models have demonstrated the possibility of islet regeneration through treatment with natural extracts, such as Allium sativum (garlic). This study aimed to investigate the effect of garlic extract (GE) on the expression of three genes (Ngn3, Pdx1, and MafA) in the pancreas and liver of diabetic rats. Thirty-two rats were divided into two groups, streptozotocin (STZ)-induced diabetic rats (n = 16) and healthy rats (n = 16). Both groups were subdivided into GE-treated (n = 8), and those administered 0.9% normal saline (NS) (n = 8) for 1 week (n = 4) and 8 weeks (n = 4). In the pancreas of diabetic rats treated with GE for 1 week, all three genes, Ngn3, Pdx1, and MafA, were significantly upregulated (p ≤ 0.01, p ≤ 0.05, and p ≤ 0.001, respectively) when compared to diabetic rats treated with NS only. However, after eight weeks of GE treatment, the expression of all three genes decreased as blood insulin increased. In the liver, only Pdx1 expression significantly (p ≤ 0.05) increased after 8 weeks. The significant expression of Ngn3, Pdx1, and MafA in the pancreas by week 1 may have induced the maturation of juvenile β-cells, which escaped the effects of STZ and caused an increase in serum insulin.
Collapse
|
21
|
Rodriguez UA, Dahiya S, Raymond ML, Gao C, Martins-Cargill CP, Piganelli JD, Gittes GK, Hu J, Esni F. Focal adhesion kinase-mediated signaling controls the onset of pancreatic cell differentiation. Development 2022; 149:dev200761. [PMID: 36017799 PMCID: PMC9482336 DOI: 10.1242/dev.200761] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 08/02/2022] [Indexed: 11/20/2022]
Abstract
Signals from the endothelium play a pivotal role in pancreatic lineage commitment. As such, the fate of the epithelial cells relies heavily on the spatiotemporal recruitment of the endothelial cells to the embryonic pancreas. Although it is known that VEGFA secreted by the epithelium recruits the endothelial cells to the specific domains within the developing pancreas, the mechanism that controls the timing of such recruitment is poorly understood. Here, we have assessed the role of focal adhesion kinase (FAK) in mouse pancreatic development based on our observation that the presence of the enzymatically active form of FAK (pFAK) in the epithelial cells is inversely correlated with vessel recruitment. To study the role of FAK in the pancreas, we conditionally deleted the gene encoding focal adhesion kinase in the developing mouse pancreas. We found that homozygous deletion of Fak (Ptk2) during embryogenesis resulted in ectopic epithelial expression of VEGFA, abnormal endothelial recruitment and a delay in endocrine and acinar cell differentiation. The heterozygous mutants were born with no pancreatic phenotype but displayed gradual acinar atrophy due to cell polarity defects in exocrine cells. Together, our findings imply a role for FAK in controlling the timing of pancreatic lineage commitment and/or differentiation in the embryonic pancreas by preventing endothelial recruitment to the embryonic pancreatic epithelium.
Collapse
Affiliation(s)
- Uylissa A. Rodriguez
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA 15244, USA
| | - Shakti Dahiya
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA 15244, USA
| | - Michelle L. Raymond
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA 15244, USA
| | - Chenxi Gao
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA 15244, USA
| | - Christina P. Martins-Cargill
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA 15244, USA
| | - Jon D. Piganelli
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA 15244, USA
| | - George K. Gittes
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA 15244, USA
| | - Jing Hu
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA 15244, USA
| | - Farzad Esni
- Department of Surgery, Division of Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA 15244, USA
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA 15244, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA 15123, USA
| |
Collapse
|
22
|
Varghese SS, Dhawan S. Polycomb Repressive Complexes: Shaping Pancreatic Beta-Cell Destiny in Development and Metabolic Disease. Front Cell Dev Biol 2022; 10:868592. [PMID: 35602600 PMCID: PMC9116887 DOI: 10.3389/fcell.2022.868592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 04/15/2022] [Indexed: 11/13/2022] Open
Abstract
Pancreatic beta-cells secrete the hormone insulin, which is essential for the regulation of systemic glucose homeostasis. Insufficiency of insulin due to loss of functional beta-cells results in diabetes. Epigenetic mechanisms orchestrate the stage-specific transcriptional programs that guide the differentiation, functional maturation, growth, and adaptation of beta-cells in response to growth and metabolic signals throughout life. Primary among these mechanisms is regulation by the Polycomb Repressive Complexes (PRC) that direct gene-expression via histone modifications. PRC dependent histone modifications are pliable and provide a degree of epigenetic plasticity to cellular processes. Their modulation dictates the spatio-temporal control of gene-expression patterns underlying beta-cell homeostasis. Emerging evidence shows that dysregulation of PRC-dependent epigenetic control is also a hallmark of beta-cell failure in diabetes. This minireview focuses on the multifaceted contributions of PRC modules in the specification and maintenance of terminally differentiated beta-cell phenotype, as well as beta-cell growth and adaptation. We discuss the interaction of PRC regulation with different signaling pathways and mechanisms that control functional beta-cell mass. We also highlight recent advances in our understanding of the epigenetic regulation of beta-cell homeostasis through the lens of beta-cell pathologies, namely diabetes and insulinomas, and the translational relevance of these findings. Using high-resolution epigenetic profiling and epigenetic engineering, future work is likely to elucidate the PRC regulome in beta-cell adaptation versus failure in response to metabolic challenges and identify opportunities for therapeutic interventions.
Collapse
|
23
|
Casamitjana J, Espinet E, Rovira M. Pancreatic Organoids for Regenerative Medicine and Cancer Research. Front Cell Dev Biol 2022; 10:886153. [PMID: 35592251 PMCID: PMC9110799 DOI: 10.3389/fcell.2022.886153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/06/2022] [Indexed: 11/16/2022] Open
Abstract
In recent years, the development of ex vivo organoid cultures has gained substantial attention as a model to study regenerative medicine and diseases in several tissues. Diabetes and pancreatic ductal adenocarcinoma (PDAC) are the two major devastating diseases affecting the pancreas. Suitable models for regenerative medicine in diabetes and to accurately study PDAC biology and treatment response are essential in the pancreatic field. Pancreatic organoids can be generated from healthy pancreas or pancreatic tumors and constitute an important translational bridge between in vitro and in vivo models. Here, we review the rapidly emerging field of pancreatic organoids and summarize the current applications of the technology to tissue regeneration, disease modelling, and drug screening.
Collapse
Affiliation(s)
- Joan Casamitjana
- Department of Physiological Science, School of Medicine, University of Barcelona (UB), L'Hospitalet de Llobregat, Barcelona, Spain
- Pancreas Regeneration: Pancreatic Progenitors and Their Niche Group, Regenerative Medicine Program, Institut D’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain
- Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P-CMR[C]), L’Hospitalet de Llobregat, Barcelona, Spain
| | - Elisa Espinet
- Department of Pathology and Experimental Therapy, School of Medicine, University of Barcelona (UB), L’Hospitalet de Llobregat, Barcelona, Spain
- Molecular Mechanisms and Experimental Therapy in Oncology Program (Oncobell), Institut D’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain
| | - Meritxell Rovira
- Department of Physiological Science, School of Medicine, University of Barcelona (UB), L'Hospitalet de Llobregat, Barcelona, Spain
- Pancreas Regeneration: Pancreatic Progenitors and Their Niche Group, Regenerative Medicine Program, Institut D’Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain
- Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P-CMR[C]), L’Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
24
|
Wang J, Wang D, Chen X, Yuan S, Bai L, Liu C, Zeng YA. Isolation of mouse pancreatic islet Procr + progenitors and long-term expansion of islet organoids in vitro. Nat Protoc 2022; 17:1359-1384. [PMID: 35396545 DOI: 10.1038/s41596-022-00683-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/22/2021] [Indexed: 12/22/2022]
Abstract
Insulin production is required for glucose homeostasis. Pancreatic islet β cells are the only cells that produce insulin in humans; however, generation of functional β cells in vitro from embryonic or adult tissues has been challenging. Here, we describe isolation of pancreatic islet progenitors from adult mice, which enables the efficient generation and long-term expansion of functional islet organoids in vitro. This protocol starts with purification of protein C receptor (Procr)-expressing islet progenitors. Coculture with endothelial cells generates islet organoids in vitro that can be expanded by passage. Functional maturation is achieved as a consequence of a prolonged culture period and cyclic glucose stimulation. Primary islet organoids form in 7-10 days. Subsequently, each passage takes 1 week, with the final maturation step requiring 3 weeks of additional culture. The resulting organoids are predominantly composed of β cells but also contain small proportions of α, δ and pancreatic polypeptide cells. The organoids sense glucose and secrete insulin. This approach thus provides a strategy for β cell generation in vitro and an organoid system to study islet regeneration and diseases.
Collapse
Affiliation(s)
- Jingqiang Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Daisong Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- Hubrecht Institute and Oncode Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre Utrecht, Netherlands, Utrecht University and Princess Maxima Center, Utrecht, Netherlands
| | - Xinyi Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shubo Yuan
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Lanyue Bai
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Chunye Liu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yi Arial Zeng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, China.
- Bio-Research Innovation Center, Institute of Biochemistry and Cell Biology, Suzhou, China.
| |
Collapse
|
25
|
Perez-Frances M, Abate MV, Baronnier D, Scherer PE, Fujitani Y, Thorel F, Herrera PL. Adult pancreatic islet endocrine cells emerge as fetal hormone-expressing cells. Cell Rep 2022; 38:110377. [PMID: 35172145 PMCID: PMC8864465 DOI: 10.1016/j.celrep.2022.110377] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 12/07/2021] [Accepted: 01/21/2022] [Indexed: 12/13/2022] Open
Abstract
The precise developmental dynamics of the pancreatic islet endocrine cell types, and their interrelation, are unknown. Some authors claim the persistence of islet cell differentiation from precursor cells after birth (“neogenesis”). Here, using four conditional cell lineage tracing (“pulse-and-chase”) murine models, we describe the natural history of pancreatic islet cells, once they express a hormone gene, until late in life. Concerning the contribution of early-appearing embryonic hormone-expressing cells to the formation of islets, we report that adult islet cells emerge from embryonic hormone-expressing cells arising at different time points during development, without any evidence of postnatal neogenesis. We observe specific patterns of hormone gene activation and switching during islet morphogenesis, revealing that, within each cell type, cells have heterogeneous developmental trajectories. This likely applies to most maturating cells in the body, and explains the observed phenotypic variability within differentiated cell types. Such knowledge should help devising novel regenerative therapies. Adult pancreatic islet endocrine cells arise as embryonic hormone-expressing cells No detectable islet cell differentiation from putative precursor cells after birth Some embryonic hormone-producing cells display a switch in hormone gene expression
Collapse
Affiliation(s)
- Marta Perez-Frances
- Department of Genetic Medicine & Development, iGE3 and Centre Facultaire du Diabète, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Maria Valentina Abate
- Department of Genetic Medicine & Development, iGE3 and Centre Facultaire du Diabète, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Delphine Baronnier
- Department of Genetic Medicine & Development, iGE3 and Centre Facultaire du Diabète, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Philipp E Scherer
- Touchstone Diabetes Center, Departments of Internal Medicine and Cell Biology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-8549, USA
| | - Yoshio Fujitani
- Laboratory of Developmental Biology & Metabolism, Institute for Molecular & Cellular Regulation, Gunma University, 3-39-15 Showa-machi, Maebashi, Gunma, 371-8512, Japan
| | - Fabrizio Thorel
- Department of Genetic Medicine & Development, iGE3 and Centre Facultaire du Diabète, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Pedro L Herrera
- Department of Genetic Medicine & Development, iGE3 and Centre Facultaire du Diabète, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland.
| |
Collapse
|
26
|
Daly CA, Hall ET, Ogden SK. Regulatory mechanisms of cytoneme-based morphogen transport. Cell Mol Life Sci 2022; 79:119. [PMID: 35119540 PMCID: PMC8816744 DOI: 10.1007/s00018-022-04148-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/05/2022] [Accepted: 01/12/2022] [Indexed: 01/07/2023]
Abstract
During development and tissue homeostasis, cells must communicate with their neighbors to ensure coordinated responses to instructional cues. Cues such as morphogens and growth factors signal at both short and long ranges in temporal- and tissue-specific manners to guide cell fate determination, provide positional information, and to activate growth and survival responses. The precise mechanisms by which such signals traverse the extracellular environment to ensure reliable delivery to their intended cellular targets are not yet clear. One model for how this occurs suggests that specialized filopodia called cytonemes extend between signal-producing and -receiving cells to function as membrane-bound highways along which information flows. A growing body of evidence supports a crucial role for cytonemes in cell-to-cell communication. Despite this, the molecular mechanisms by which cytonemes are initiated, how they grow, and how they deliver specific signals are only starting to be revealed. Herein, we discuss recent advances toward improved understanding of cytoneme biology. We discuss similarities and differences between cytonemes and other types of cellular extensions, summarize what is known about how they originate, and discuss molecular mechanisms by which their activity may be controlled in development and tissue homeostasis. We conclude by highlighting important open questions regarding cytoneme biology, and comment on how a clear understanding of their function may provide opportunities for treating or preventing disease.
Collapse
Affiliation(s)
- Christina A Daly
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Pl. MS340, Memphis, TN, 38105, USA
- St. Jude Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Pl, MS 1500, Memphis, TN, 38105, USA
| | - Eric T Hall
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Pl. MS340, Memphis, TN, 38105, USA
| | - Stacey K Ogden
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Pl. MS340, Memphis, TN, 38105, USA.
| |
Collapse
|
27
|
Glorieux L, Sapala A, Willnow D, Moulis M, Salowka A, Darrigrand JF, Edri S, Schonblum A, Sakhneny L, Schaumann L, Gómez HF, Lang C, Conrad L, Guillemot F, Levenberg S, Landsman L, Iber D, Pierreux CE, Spagnoli FM. Development of a 3D atlas of the embryonic pancreas for topological and quantitative analysis of heterologous cell interactions. Development 2022; 149:274013. [PMID: 35037942 PMCID: PMC8918780 DOI: 10.1242/dev.199655] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 12/20/2021] [Indexed: 01/05/2023]
Abstract
Generating comprehensive image maps, while preserving spatial three-dimensional (3D) context, is essential in order to locate and assess quantitatively specific cellular features and cell-cell interactions during organ development. Despite recent advances in 3D imaging approaches, our current knowledge of the spatial organization of distinct cell types in the embryonic pancreatic tissue is still largely based on two-dimensional histological sections. Here, we present a light-sheet fluorescence microscopy approach to image the pancreas in three dimensions and map tissue interactions at key time points in the mouse embryo. We demonstrate the utility of the approach by providing volumetric data, 3D distribution of three main cellular components (epithelial, mesenchymal and endothelial cells) within the developing pancreas, and quantification of their relative cellular abundance within the tissue. Interestingly, our 3D images show that endocrine cells are constantly and increasingly in contact with endothelial cells forming small vessels, whereas the interactions with mesenchymal cells decrease over time. These findings suggest distinct cell-cell interaction requirements for early endocrine cell specification and late differentiation. Lastly, we combine our image data in an open-source online repository (referred to as the Pancreas Embryonic Cell Atlas). Summary: A light-sheet fluorescence microscopy approach is used for 3D imaging of the pancreas and to quantitatively map its interactions with surrounding tissues at key development time points in the mouse embryo.
Collapse
Affiliation(s)
- Laura Glorieux
- Cell Biology Unit, de Duve Institute, UCLouvain, Woluwe 1200, Belgium
| | - Aleksandra Sapala
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Basel 4058, Switzerland.,Swiss Institute of Bioinformatics (SIB), Basel 4058, Switzerland
| | - David Willnow
- Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Manon Moulis
- Cell Biology Unit, de Duve Institute, UCLouvain, Woluwe 1200, Belgium
| | - Anna Salowka
- Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Jean-Francois Darrigrand
- Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Shlomit Edri
- Department of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Anat Schonblum
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Lina Sakhneny
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Laura Schaumann
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Basel 4058, Switzerland.,Swiss Institute of Bioinformatics (SIB), Basel 4058, Switzerland
| | - Harold F Gómez
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Basel 4058, Switzerland.,Swiss Institute of Bioinformatics (SIB), Basel 4058, Switzerland
| | - Christine Lang
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Basel 4058, Switzerland.,Swiss Institute of Bioinformatics (SIB), Basel 4058, Switzerland
| | - Lisa Conrad
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Basel 4058, Switzerland.,Swiss Institute of Bioinformatics (SIB), Basel 4058, Switzerland
| | | | - Shulamit Levenberg
- Department of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Limor Landsman
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Dagmar Iber
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Basel 4058, Switzerland.,Swiss Institute of Bioinformatics (SIB), Basel 4058, Switzerland
| | | | - Francesca M Spagnoli
- Centre for Stem Cell and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| |
Collapse
|
28
|
Vokurka M, Lacina L, Brábek J, Kolář M, Ng YZ, Smetana K. Cancer-Associated Fibroblasts Influence the Biological Properties of Malignant Tumours via Paracrine Secretion and Exosome Production. Int J Mol Sci 2022; 23:964. [PMID: 35055153 PMCID: PMC8778626 DOI: 10.3390/ijms23020964] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/10/2022] [Accepted: 01/13/2022] [Indexed: 12/15/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs) are an essential component of the tumour microenvironment. They represent a heterogeneous group of cells that are under the control of cancer cells and can reversely influence the cancer cell population. They affect the cancer cell differentiation status, and the migration and formation of metastases. This is achieved through the production of the extracellular matrix and numerous bioactive factors. IL-6 seems to play the central role in the communication of noncancerous and cancer cells in the tumour. This review outlines the role of exosomes in cancer cells and cancer-associated fibroblasts. Available data on the exosomal cargo, which can significantly intensify interactions in the tumour, are summarised. The role of exosomes as mediators of the dialogue between cancer cells and cancer-associated fibroblasts is discussed together with their therapeutic relevance. The functional unity of the paracrine- and exosome-mediated communication of cancer cells with the tumour microenvironment represented by CAFs is worthy of attention.
Collapse
Affiliation(s)
- Martin Vokurka
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, 120 00 Prague 2, Czech Republic;
| | - Lukáš Lacina
- Institute of Anatomy, First Faculty of Medicine, Charles University, 120 00 Prague 2, Czech Republic
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department of Dermatovenereology, First Faculty of Medicine, Charles University and General University Hospital, 120 00 Prague, Czech Republic
| | - Jan Brábek
- Department of Cell Biology, Faculty of Science, Charles University, 120 00 Prague 2, Czech Republic;
- BIOCEV, Faculty of Science, Charles University, 252 50 Vestec, Czech Republic
| | - Michal Kolář
- Institute of Molecular Genetics, Czech Academy of Sciences, 142 20 Prague 4, Czech Republic;
| | - Yi Zhen Ng
- A*STAR Skin Research Labs (A*SRL)—Biopolis, Skin Research Institute of Singapore, 8A Biomedical Grove #06-06 Immunos Singapore, Singapore 138665, Singapore;
| | - Karel Smetana
- Institute of Anatomy, First Faculty of Medicine, Charles University, 120 00 Prague 2, Czech Republic
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
| |
Collapse
|
29
|
Mehta V, Hopson PE, Smadi Y, Patel SB, Horvath K, Mehta DI. Development of the human pancreas and its exocrine function. Front Pediatr 2022; 10:909648. [PMID: 36245741 PMCID: PMC9557127 DOI: 10.3389/fped.2022.909648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 08/11/2022] [Indexed: 11/25/2022] Open
Abstract
The pancreas has both endocrine and exocrine function and plays an important role in digestion and glucose control. Understanding the development of the pancreas, grossly and microscopically, and the genetic factors regulating it provides further insight into clinical problems that arise when these processes fail. Animal models of development are known to have inherent issues when understanding human development. Therefore, in this review, we focus on human studies that have reported gross and microscopic development including acinar-, ductal-, and endocrine cells and the neural network. We review the genes and transcription factors involved in organ formation using data from animal models to bridge current understanding where necessary. We describe the development of exocrine function in the fetus and postnatally. A deeper review of the genes involved in pancreatic formation allows us to describe the development of the different groups (proteases, lipids, and amylase) of enzymes during fetal life and postnatally and describe the genetic defects. We discuss the constellation of gross anatomical, as well as microscopic defects that with genetic mutations lead to pancreatic insufficiency and disease states.
Collapse
Affiliation(s)
- Vijay Mehta
- Center for Digestive Health and Nutrition, Arnold Palmer Hospital for Children, Orlando, FL, United States
| | - Puanani E Hopson
- Department of Children Center, Pediatric and Adolescent Medicine, Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States
| | - Yamen Smadi
- Center for Digestive Health and Nutrition, Arnold Palmer Hospital for Children, Orlando, FL, United States
| | - Samit B Patel
- Pediatric Gastroenterology and Nutrition of Tampa Bay, Tampa Bay, FL, United States
| | - Karoly Horvath
- Center for Digestive Health and Nutrition, Arnold Palmer Hospital for Children, Orlando, FL, United States
| | - Devendra I Mehta
- Center for Digestive Health and Nutrition, Arnold Palmer Hospital for Children, Orlando, FL, United States
| |
Collapse
|
30
|
van Roey R, Brabletz T, Stemmler MP, Armstark I. Deregulation of Transcription Factor Networks Driving Cell Plasticity and Metastasis in Pancreatic Cancer. Front Cell Dev Biol 2021; 9:753456. [PMID: 34888306 PMCID: PMC8650502 DOI: 10.3389/fcell.2021.753456] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/27/2021] [Indexed: 12/15/2022] Open
Abstract
Pancreatic cancer is a very aggressive disease with 5-year survival rates of less than 10%. The constantly increasing incidence and stagnant patient outcomes despite changes in treatment regimens emphasize the requirement of a better understanding of the disease mechanisms. Challenges in treating pancreatic cancer include diagnosis at already progressed disease states due to the lack of early detection methods, rapid acquisition of therapy resistance, and high metastatic competence. Pancreatic ductal adenocarcinoma, the most prevalent type of pancreatic cancer, frequently shows dominant-active mutations in KRAS and TP53 as well as inactivation of genes involved in differentiation and cell-cycle regulation (e.g. SMAD4 and CDKN2A). Besides somatic mutations, deregulated transcription factor activities strongly contribute to disease progression. Specifically, transcriptional regulatory networks essential for proper lineage specification and differentiation during pancreas development are reactivated or become deregulated in the context of cancer and exacerbate progression towards an aggressive phenotype. This review summarizes the recent literature on transcription factor networks and epigenetic gene regulation that play a crucial role during tumorigenesis.
Collapse
Affiliation(s)
- Ruthger van Roey
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Marc P Stemmler
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Isabell Armstark
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
31
|
Scoville DW, Jetten AM. GLIS3: A Critical Transcription Factor in Islet β-Cell Generation. Cells 2021; 10:cells10123471. [PMID: 34943978 PMCID: PMC8700524 DOI: 10.3390/cells10123471] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/23/2021] [Accepted: 12/06/2021] [Indexed: 11/22/2022] Open
Abstract
Understanding of pancreatic islet biology has greatly increased over the past few decades based in part on an increased understanding of the transcription factors that guide this process. One such transcription factor that has been increasingly tied to both β-cell development and the development of diabetes in humans is GLIS3. Genetic deletion of GLIS3 in mice and humans induces neonatal diabetes, while single nucleotide polymorphisms (SNPs) in GLIS3 have been associated with both Type 1 and Type 2 diabetes. As a significant progress has been made in understanding some of GLIS3’s roles in pancreas development and diabetes, we sought to compare current knowledge on GLIS3 within the pancreas to that of other islet enriched transcription factors. While GLIS3 appears to regulate similar genes and pathways to other transcription factors, its unique roles in β-cell development and maturation make it a key target for future studies and therapy.
Collapse
|
32
|
Backx E, Coolens K, Van den Bossche JL, Houbracken I, Espinet E, Rooman I. On the Origin of Pancreatic Cancer: Molecular Tumor Subtypes in Perspective of Exocrine Cell Plasticity. Cell Mol Gastroenterol Hepatol 2021; 13:1243-1253. [PMID: 34875393 PMCID: PMC8881661 DOI: 10.1016/j.jcmgh.2021.11.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/30/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating type of cancer. While many studies have shed light into the pathobiology of PDAC, the nature of PDAC's cell of origin remains under debate. Studies in adult pancreatic tissue have unveiled a remarkable exocrine cell plasticity including transitional states, mostly exemplified by acinar to ductal cell metaplasia, but also with recent evidence hinting at duct to basal cell transitions. Single-cell RNA sequencing has further revealed intrapopulation heterogeneity among acinar and duct cells. Transcriptomic and epigenomic relationships between these exocrine cell differentiation states and PDAC molecular subtypes have started to emerge, suggesting different ontogenies for different tumor subtypes. This review sheds light on these diverse aspects with particular focus on studies with human cells. Understanding the "masked ball" of exocrine cells at origin of PDAC and leaving behind the binary acinar vs duct cell classification may significantly advance our insights in PDAC biology.
Collapse
Affiliation(s)
- Elyne Backx
- Laboratory of Medical and Molecular Oncology, Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Katarina Coolens
- Laboratory of Medical and Molecular Oncology, Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jan-Lars Van den Bossche
- Laboratory of Medical and Molecular Oncology, Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Isabelle Houbracken
- Laboratory of Medical and Molecular Oncology, Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Elisa Espinet
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine, Heidelberg, Germany; Division of Stem Cells and Cancer, German Cancer Research Center, Heidelberg, Germany
| | - Ilse Rooman
- Laboratory of Medical and Molecular Oncology, Oncology Research Center, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
33
|
Wang K, Deng Y, Zhang J, Cheng B, Huang Y, Meng Y, Zhong K, Xiong G, Guo J, Liu Y, Lu H. Toxicity of thioacetamide and protective effects of quercetin in zebrafish (Danio rerio) larvae. ENVIRONMENTAL TOXICOLOGY 2021; 36:2062-2072. [PMID: 34227734 DOI: 10.1002/tox.23323] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 06/17/2021] [Accepted: 06/27/2021] [Indexed: 06/13/2023]
Abstract
Quercetin is a flavonoid compound with a variety of biological properties that is widely distributed throughout the plant kingdom. Studies have found that quercetin has anti-inflammatory, antioxidant, and liver-protective effects, while thioacetamide (TAA) can cause inflammation and liver damage in zebrafish larvae. The purpose of this study was to evaluate whether quercetin can prevent TAA-induced inflammation and liver damage in zebrafish larvae and to investigate the molecular mechanisms involved. Zebrafish Tg transgenic lines were used as the experimental animals. Behavioral, oxidative stress level, proliferative antigen chromogenic antibody, and western blot analyses were carried out on zebrafish larvae in the control group and groups treated with TAA and 12 μM quercetin. The results indicated that quercetin promoted the development of zebrafish larvae damaged by TAA, exhibited antioxidant and anti-inflammatory properties, and promoted cell proliferation. Quercetin reduced the expression of p53 protein in zebrafish larvae injured by TAA, resulting in decreased levels of Bax and increased levels of Bcl-2. The findings suggested quercetin has antiapoptotic action. Quercetin reduced the expression of DKK1 and DKK2 genes related to the Wnt signaling pathway in zebrafish larvae damaged by TAA and increased the expression of Lef1 and wnt2bb. Quercetin may regulate the development of zebrafish larvae damaged by TAA through the Wnt signaling pathway. This study provides the scientific basis for the development and utilization of quercetin and the development of new related drugs.
Collapse
Affiliation(s)
- Kexin Wang
- College of life sciences, Jiangxi Normal university, Nanchang, Jiangxi, China
| | - Yunyun Deng
- College of life sciences, Jiangxi Normal university, Nanchang, Jiangxi, China
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, Jiangxi, China
- Jiangxi Key Laboratory of Developmental Biology of Organs, Ji'an, Jiangxi, China
| | - June Zhang
- College of life sciences, Jiangxi Normal university, Nanchang, Jiangxi, China
| | - Bo Cheng
- Center for drug screening and research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, Jiangxi, China
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, Jiangxi, China
- Jiangxi Key Laboratory of Developmental Biology of Organs, Ji'an, Jiangxi, China
| | - Yong Huang
- Center for drug screening and research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, Jiangxi, China
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, Jiangxi, China
- Jiangxi Key Laboratory of Developmental Biology of Organs, Ji'an, Jiangxi, China
| | - Yunlong Meng
- Center for drug screening and research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, Jiangxi, China
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, Jiangxi, China
- Jiangxi Key Laboratory of Developmental Biology of Organs, Ji'an, Jiangxi, China
| | - Keyuan Zhong
- Center for drug screening and research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, Jiangxi, China
| | - Guanghua Xiong
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, Jiangxi, China
- Jiangxi Key Laboratory of Developmental Biology of Organs, Ji'an, Jiangxi, China
| | - Jing Guo
- College of life sciences, Jiangxi Normal university, Nanchang, Jiangxi, China
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, Jiangxi, China
- Jiangxi Key Laboratory of Developmental Biology of Organs, Ji'an, Jiangxi, China
| | - Yi Liu
- College of life sciences, Jiangxi Normal university, Nanchang, Jiangxi, China
| | - Huiqiang Lu
- Center for drug screening and research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, Jiangxi, China
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, Jiangxi, China
- Jiangxi Key Laboratory of Developmental Biology of Organs, Ji'an, Jiangxi, China
| |
Collapse
|
34
|
Li J, Chen B, Fellows GF, Goodyer CG, Wang R. Activation of Pancreatic Stellate Cells Is Beneficial for Exocrine but Not Endocrine Cell Differentiation in the Developing Human Pancreas. Front Cell Dev Biol 2021; 9:694276. [PMID: 34490247 PMCID: PMC8418189 DOI: 10.3389/fcell.2021.694276] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/14/2021] [Indexed: 02/04/2023] Open
Abstract
Pancreatic stellate cells (PaSCs) are non-endocrine, mesenchymal-like cells that reside within the peri-pancreatic tissue of the rodent and human pancreas. PaSCs regulate extracellular matrix (ECM) turnover in maintaining the integrity of pancreatic tissue architecture. Although there is evidence indicating that PaSCs are involved in islet cell survival and function, its role in islet cell differentiation during human pancreatic development remains unclear. The present study examines the expression pattern and functional role of PaSCs in islet cell differentiation of the developing human pancreas from late 1st to 2nd trimester of pregnancy. The presence of PaSCs in human pancreata (8–22 weeks of fetal age) was characterized by ultrastructural, immunohistological, quantitative RT-PCR and western blotting approaches. Using human fetal PaSCs derived from pancreata at 14–16 weeks, freshly isolated human fetal islet-epithelial cell clusters (hIECCs) were co-cultured with active or inactive PaSCs in vitro. Ultrastructural and immunofluorescence analysis demonstrated a population of PaSCs near ducts and newly formed islets that appeared to make complex cell-cell dendritic-like contacts. A small subset of PaSCs co-localized with pancreatic progenitor-associated transcription factors (PDX1, SOX9, and NKX6-1). PaSCs were highly proliferative, with significantly higher mRNA and protein levels of PaSC markers (desmin, αSMA) during the 1st trimester of pregnancy compared to the 2nd trimester. Isolated human fetal PaSCs were identified by expression of stellate cell markers and ECM. Suppression of PaSC activation, using all-trans retinoic acid (ATRA), resulted in reduced PaSC proliferation and ECM proteins. Co-culture of hIECCs, directly on PaSCs or indirectly using Millicell® Inserts or using PaSC-conditioned medium, resulted in a reduction the number of insulin+ cells but a significant increase in the number of amylase+ cells. Suppression of PaSC activation or Notch activity during the co-culture resulted in an increase in beta-cell differentiation. This study determined that PaSCs, abundant during the 1st trimester of pancreatic development but decreased in the 2nd trimester, are located near ductal and islet structures. Direct and indirect co-cultures of hIECCs with PaSCs suggest that activation of PaSCs has opposing effects on beta-cell and exocrine cell differentiation during human fetal pancreas development, and that these effects may be dependent on Notch signaling.
Collapse
Affiliation(s)
- Jinming Li
- Children's Health Research Institute, Western University, London, ON, Canada.,Departments of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Bijun Chen
- Children's Health Research Institute, Western University, London, ON, Canada
| | - George F Fellows
- Department of Obstetrics and Gynecology, Western University, London, ON, Canada
| | | | - Rennian Wang
- Children's Health Research Institute, Western University, London, ON, Canada.,Departments of Physiology and Pharmacology, Western University, London, ON, Canada
| |
Collapse
|
35
|
Affiliation(s)
- Jason R Pitarresi
- Division of Gastroenterology, Department of Medicine, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Division of Digestive and Liver Diseases, Department of Medicine, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Anil K Rustgi
- Division of Digestive and Liver Diseases, Department of Medicine, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
36
|
Maternal Under- and Over-Nutrition during Gestation Causes Islet Hypertrophy and Sex-Specific Changes to Pancreas DNA Methylation in Fetal Sheep. Animals (Basel) 2021; 11:ani11092531. [PMID: 34573497 PMCID: PMC8466738 DOI: 10.3390/ani11092531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 01/18/2023] Open
Abstract
The mechanisms by which fetal programming predisposes offspring to reduced β-cell function later in life are poorly understood. We hypothesized that maternal under- and over-nutrition during gestation would negatively affect offspring pancreas development and alter DNA methylation patterns. Pregnant ewes (n = 78) were fed 100, 60, or 140% of NRC requirements beginning at d 30.2 ± 0.2 of gestation. The fetuses are referred to as CON, RES, and OVER, respectively. Fetal pancreas tissue was collected at d 90 or 135 of gestation or within 24 h of birth. Tissue was preserved for histological (n = 8 to 9 offspring per treatment per time point) and DNA methylation analyses (n = 3 to 4 fetuses per treatment per sex). At d 135, OVER exhibited an increased islet size, reduced islet number, and greater insulin positive area compared with CON (p ≤ 0.03). An increased islet size was also observed at d 135 in RES (p ≤ 0.03) compared with CON. Cellular proliferation was reduced at birth in OVER vs. CON (p = 0.01). In the RES vs. CON females, 62% of the differentially methylated regions (DMRs) were hypomethylated (p ≤ 0.001). In the RES vs. CON males, 93% of the DMRs were hypermethylated (p ≤ 0.001). In OVER, 66 and 80% of the DMRs were hypermethylated in the female and male offspring compared with CON (p ≤ 0.001). In conclusion, changes to maternal diet during pregnancy affects the islet hypertrophy and cellular proliferation of the offspring at early post-natal time points. Additionally, changes in DNA methylation patterns appear to be in a diet-specific and sex-dependent manner.
Collapse
|
37
|
Chitosan/Gelatin/PVA Scaffolds for Beta Pancreatic Cell Culture. Polymers (Basel) 2021; 13:polym13142372. [PMID: 34301129 PMCID: PMC8309518 DOI: 10.3390/polym13142372] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 07/11/2021] [Accepted: 07/13/2021] [Indexed: 02/07/2023] Open
Abstract
Chitosan scaffolds based on blending polymers are a common strategy used in tissue engineering. The objective of this study was evaluation the properties of scaffolds based on a ternary blend of chitosan (Chi), gelatin (Ge), and polyvinyl alcohol (PVA) (Chi/Ge/PVA), which were prepared by cycles of freeze-thawing and freeze-drying. It then was used for three-dimensional BRIN-BD11 beta-cells culturing. Weight ratios of Chi/Ge/PVA (1:1:1, 2:2:1, 2:3:1, and 3:2:1) were proposed and porosity, pore size, degradation, swelling rate, compressive strength, and cell viability analyzed. All ternary blend scaffolds structures are highly porous (with a porosity higher than 80%) and interconnected. The pore size distribution varied from 0.6 to 265 μm. Ternary blends scaffolds had controllable degradation rates compared to binary blend scaffolds, and an improved swelling capacity of the samples with increasing chitosan concentration was found. An increase in Young’s modulus and compressive strength was observed with increasing gelatin concentration. The highest compressive strength reached 101.6 Pa. The MTT assay showed that the ternary blends scaffolds P3 and P4 supported cell viability better than the binary blend scaffold. Therefore, these results illustrated that ternary blends scaffolds P3 and P4 could provide a better environment for BRIN-BD11 cell proliferation.
Collapse
|
38
|
Architecture of the Pancreatic Islets and Endocrine Cell Arrangement in the Embryonic Pancreas of the Grass Snake ( Natrix natrix L.). Immunocytochemical Studies and 3D Reconstructions. Int J Mol Sci 2021; 22:ijms22147601. [PMID: 34299221 PMCID: PMC8304277 DOI: 10.3390/ijms22147601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 11/17/2022] Open
Abstract
During the early developmental stages of grass snakes, within the differentiating pancreas, cords of endocrine cells are formed. They differentiate into agglomerates of large islets flanked throughout subsequent developmental stages by small groups of endocrine cells forming islets. The islets are located within the cephalic part of the dorsal pancreas. At the end of the embryonic period, the pancreatic islet agglomerates branch off, and as a result of their remodeling, surround the splenic "bulb". The stage of pancreatic endocrine ring formation is the first step in formation of intrasplenic islets characteristics for the adult specimens of the grass snake. The arrangement of endocrine cells within islets changes during pancreas differentiation. Initially, the core of islets formed from B and D cells is surrounded by a cluster of A cells. Subsequently, A, B, and D endocrine cells are mixed throughout the islets. Before grass snake hatching, A and B endocrine cells are intermingled within the islets, but D cells are arranged centrally. Moreover, the pancreatic polypeptide (PP) cells are not found within the embryonic pancreas of the grass snake. Variation in the proportions of different cell types, depending on the part of the pancreas, may affect the islet function-a higher proportion of glucagon cells is beneficial for insulin secretion.
Collapse
|
39
|
Milan M, Diaferia GR, Natoli G. Tumor cell heterogeneity and its transcriptional bases in pancreatic cancer: a tale of two cell types and their many variants. EMBO J 2021; 40:e107206. [PMID: 33844319 PMCID: PMC8246061 DOI: 10.15252/embj.2020107206] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 12/14/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), one of the most highly lethal tumors, is characterized by complex histology, with a massive fibrotic stroma in which both pseudo-glandular structures and compact nests of abnormally differentiated tumor cells are embedded, in different proportions and with different mutual relationships in space. This complexity and the heterogeneity of the tumor component have hindered the development of a broadly accepted, clinically actionable classification of PDACs, either on a morphological or a molecular basis. Here, we discuss evidence suggesting that such heterogeneity can to a large extent, albeit not exclusively, be traced back to two main classes of PDAC cells that commonly coexist in the same tumor: cells that maintained their ability to differentiate toward endodermal, mucin-producing epithelia and epithelial cells unable to form glandular structures and instead characterized by various levels of squamous differentiation and the expression of mesenchymal lineage genes. The underlying gene regulatory networks and how they are controlled by distinct transcription factors, as well as the practical implications of these two different populations of tumor cells, are discussed.
Collapse
Affiliation(s)
- Marta Milan
- Department of Experimental OncologyEuropean Institute of Oncology (IEO) IRCCSMilanItaly
- Present address:
The Francis Crick InstituteLondonUK
| | - Giuseppe R Diaferia
- Department of Experimental OncologyEuropean Institute of Oncology (IEO) IRCCSMilanItaly
| | - Gioacchino Natoli
- Department of Experimental OncologyEuropean Institute of Oncology (IEO) IRCCSMilanItaly
- Humanitas UniversityMilanItaly
| |
Collapse
|
40
|
Baikenova MB, Chereshnev VA, Sokolova KV, Gette IF, Emelyanov VV, Danilova IG. Hepatic Insulin-Positive Cells and Major Transcription Factors (PDX1, MAFA, NGN3) in Rat Models of Type 1 and Type 2 Diabetes Mellitus. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s0022093021040037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
41
|
REST Inhibits Direct Reprogramming of Pancreatic Exocrine to Endocrine Cells by Preventing PDX1-Mediated Activation of Endocrine Genes. Cell Rep 2021; 31:107591. [PMID: 32375045 DOI: 10.1016/j.celrep.2020.107591] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 01/29/2020] [Accepted: 04/09/2020] [Indexed: 12/13/2022] Open
Abstract
The emerging appreciation of plasticity among pancreatic lineages has created interest in harnessing cellular reprogramming for β cell replacement therapy of diabetes. Current reprogramming methodologies are inefficient, largely because of a limited understanding of the underlying mechanisms. Using an in vitro reprogramming system, we reveal the transcriptional repressor RE-1 silencing transcription factor (REST) as a barrier for β cell gene expression in the reprogramming of pancreatic exocrine cells. We observe that REST-bound loci lie adjacent to the binding sites of multiple key β cell transcription factors, including PDX1. Accordingly, a loss of REST function combined with PDX1 expression results in the synergistic activation of endocrine genes. This is accompanied by increased histone acetylation and PDX1 binding at endocrine gene loci. Collectively, our data identify a mechanism for REST activity involving the prevention of PDX1-mediated activation of endocrine genes and uncover REST downregulation and the resulting chromatin alterations as key events in β cell reprogramming.
Collapse
|
42
|
Isaacson A, Spagnoli FM. Pancreatic cell fate specification: insights into developmental mechanisms and their application for lineage reprogramming. Curr Opin Genet Dev 2021; 70:32-39. [PMID: 34062490 DOI: 10.1016/j.gde.2021.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/01/2021] [Accepted: 05/02/2021] [Indexed: 12/15/2022]
Abstract
Diabetes is a group of metabolic disorders, which results from insufficient functional pancreatic β-cell mass either due to the autoimmune destruction of insulin producing β-cells, or their death or de-differentiation as compensation for insulin resistance. The ability to reprogram cell types within close developmental proximity to β-cells offers a strategy to replenish β-cell mass and a future possible treatment of diabetes. Here, we review recent advances in the fields of pancreas development and lineage reprogramming. We also probe the possibility of using reprogrammed cells as an approach by which to further understand developmental mechanisms, in particular roadblocks to changing cell identity. Finally, we highlight fundamental challenges that need to be overcome to advance lineage reprogramming for generating pancreatic cells.
Collapse
Affiliation(s)
- Abigail Isaacson
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, Floor 28, Tower Wing, Great Maze Pond, London SE1 9RT, UK
| | - Francesca M Spagnoli
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, Floor 28, Tower Wing, Great Maze Pond, London SE1 9RT, UK.
| |
Collapse
|
43
|
Cilvik SN, Wesolowski SR, Anthony RV, Brown LD, Rozance PJ. Late gestation fetal hyperglucagonaemia impairs placental function and results in diminished fetal protein accretion and decreased fetal growth. J Physiol 2021; 599:3403-3427. [PMID: 33878802 DOI: 10.1113/jp281288] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 04/06/2021] [Indexed: 12/14/2022] Open
Abstract
KEY POINTS Fetal glucagon concentrations are elevated in the setting of placental insufficiency, hypoxia and elevated stress hormones. Chronically elevated glucagon concentrations in the adult result in profound decreases in amino acid concentrations and lean body mass. Experimental elevation of fetal glucagon concentrations in a late-gestation pregnant sheep results in lower fetal amino acid concentrations, lower protein accretion and lower fetal weight, in addition to decreased placental function. This study demonstrates a negative effect of glucagon on fetal protein accretion and growth, and also provides the first example of a fetal hormone that negatively regulates placental nutrient transport and blood flow. ABSTRACT Fetal glucagon concentrations are elevated in the setting of placental insufficiency and fetal stress. Postnatal studies have demonstrated the importance of glucagon in amino acid metabolism, and limited fetal studies have suggested that glucagon inhibits umbilical uptake of certain amino acids. We hypothesized that chronic fetal hyperglucagonaemia would decrease amino acid transfer and increase amino acid oxidation by the fetus. Late gestation singleton fetal sheep received a direct intravenous infusion of glucagon (GCG; 5 or 50 ng/kg/min; n = 7 and 5, respectively) or a vehicle control (n = 10) for 8-10 days. Fetal and maternal nutrient concentrations, uterine and umbilical blood flows, fetal leucine flux, nutrient uptake rates, placental secretion of chorionic somatomammotropin (CSH), and targeted placental gene expression were measured. GCG fetuses had 13% lower fetal weight compared to controls (P = 0.0239) and >28% lower concentrations of 16 out of 21 amino acids (P < 0.02). Additionally, protein synthesis was 49% lower (P = 0.0005), and protein accretion was 92% lower in GCG fetuses (P = 0.0006). Uterine blood flow was 33% lower in ewes with GCG fetuses (P = 0.0154), while umbilical blood flow was similar. Fetal hyperglucagonaemia lowered uterine uptake of 10 amino acids by >48% (P < 0.05) and umbilical uptake of seven amino acids by >29% (P < 0.04). Placental secretion of CSH into maternal circulation was reduced by 80% compared to controls (P = 0.0080). This study demonstrates a negative effect of glucagon on fetal protein accretion and growth. It also demonstrates that glucagon, a hormone of fetal origin, negatively regulates maternal placental nutrient transport function, placental CSH production and uterine blood flow.
Collapse
Affiliation(s)
- Sarah N Cilvik
- Wake Forest University Health Sciences, Winston-Salem, NC, 27157, USA
| | | | - Russ V Anthony
- Colorado State University College of Veterinary Medicine, Fort Collins, CO, USA
| | - Laura D Brown
- University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Paul J Rozance
- University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
44
|
Parveen N, Dhawan S. DNA Methylation Patterning and the Regulation of Beta Cell Homeostasis. Front Endocrinol (Lausanne) 2021; 12:651258. [PMID: 34025578 PMCID: PMC8137853 DOI: 10.3389/fendo.2021.651258] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 04/21/2021] [Indexed: 12/14/2022] Open
Abstract
Pancreatic beta cells play a central role in regulating glucose homeostasis by secreting the hormone insulin. Failure of beta cells due to reduced function and mass and the resulting insulin insufficiency can drive the dysregulation of glycemic control, causing diabetes. Epigenetic regulation by DNA methylation is central to shaping the gene expression patterns that define the fully functional beta cell phenotype and regulate beta cell growth. Establishment of stage-specific DNA methylation guides beta cell differentiation during fetal development, while faithful restoration of these signatures during DNA replication ensures the maintenance of beta cell identity and function in postnatal life. Lineage-specific transcription factor networks interact with methylated DNA at specific genomic regions to enhance the regulatory specificity and ensure the stability of gene expression patterns. Recent genome-wide DNA methylation profiling studies comparing islets from diabetic and non-diabetic human subjects demonstrate the perturbation of beta cell DNA methylation patterns, corresponding to the dysregulation of gene expression associated with mature beta cell state in diabetes. This article will discuss the molecular underpinnings of shaping the islet DNA methylation landscape, its mechanistic role in the specification and maintenance of the functional beta cell phenotype, and its dysregulation in diabetes. We will also review recent advances in utilizing beta cell specific DNA methylation patterns for the development of biomarkers for diabetes, and targeting DNA methylation to develop translational approaches for supplementing the functional beta cell mass deficit in diabetes.
Collapse
Affiliation(s)
| | - Sangeeta Dhawan
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, United States
| |
Collapse
|
45
|
Ravi PK, Singh SR, Mishra PR. Redefining the tail of pancreas based on the islets microarchitecture and inter-islet distance: An immunohistochemical study. Medicine (Baltimore) 2021; 100:e25642. [PMID: 33907122 PMCID: PMC8084047 DOI: 10.1097/md.0000000000025642] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 03/17/2021] [Accepted: 03/20/2021] [Indexed: 11/25/2022] Open
Abstract
ABSTRACT Researchers divided the pancreas distal to the neck into 2 equal parts as the body and tail region by an arbitrary line. Surgeons considered the part of the pancreas, left to the aorta as the tail region. We performed this study to identify the transition zone of low-density to high-density islet cells for redefining the tail region.We quantified islets area proportion, beta-cell area proportion, and inter-islet distance in 9 Indian-adult-human non-diabetic pancreases from autopsy by using anti-synaptophysin and anti-insulin antibodies. Data were categorized under 3 regions like the proximal body, distal body, and distal part of the pancreas.Islet and beta-cell area proportion are progressively increased from head to tail region of the pancreas with a significant reduction in inter-islet distance and beta-cell percentage distal to the aorta. There is no significant difference in inter-islet distance and beta-cell percentage of the distal part of the body and tail region.Crowding of islets with intermingled microarchitecture begins in the pancreas distal to the aorta, which may be the beginning of the actual tail region. This study will provide insight into the preservation of islets-rich part of the pancreas during pancreatectomy and future prediction of new-onset diabetes.
Collapse
Affiliation(s)
- Praveen Kumar Ravi
- Department of Anatomy, Trichy SRM Medical College Hospital & Research Centre, Tiruchirappalli, Tamil Nadu
| | | | - Pravash Ranjan Mishra
- Department of Anatomy, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| |
Collapse
|
46
|
A Rare Case of Dorsal Agenesis of Pancreas, Choledochal Cyst, and Hirschsprung Disease in a Young Patient. ACG Case Rep J 2021; 8:e00561. [PMID: 33928176 PMCID: PMC8078483 DOI: 10.14309/crj.0000000000000561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 10/12/2020] [Indexed: 11/22/2022] Open
Abstract
Dorsal pancreatic agenesis is a rare congenital pancreatic malformation. There is just 1 reported case associating it with choledochal cyst. However, no cases have reported yet with both coexisting with Hirschsprung disease. We report a case of a 23-year-old man, presenting with on and off epigastric pain, sometimes radiating to the back. His medical records showed he had Hirschsprung disease as a neonate, for which he underwent Duhamel procedure. Ultrasound imaging revealed a choledochal cyst and a nonvisualized distal portion of the pancreas. Further cross-sectional imaging confirmed the findings—a type 1 choledochal cyst and a dorsal agenesis of the pancreas in a patient with Hirschsprung disease.
Collapse
|
47
|
Liu Q, Jiang Y, Zhu L, Qian J, Wang C, Yang T, Prasadan K, Gittes GK, Xiao X. Insulin-positive ductal cells do not migrate into preexisting islets during pregnancy. Exp Mol Med 2021; 53:605-614. [PMID: 33820959 PMCID: PMC8102600 DOI: 10.1038/s12276-021-00593-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 12/13/2020] [Accepted: 02/16/2021] [Indexed: 12/16/2022] Open
Abstract
The adult pancreatic ductal system was suggested to harbor facultative beta-cell progenitors similar to the embryonic pancreas, and the appearance of insulin-positive duct cells has been used as evidence for natural duct-to-beta-cell reprogramming. Nevertheless, the phenotype and fate of these insulin-positive cells in ducts have not been determined. Here, we used a cell-tagging dye, CFDA-SE, to permanently label pancreatic duct cells through an intraductal infusion technique. Representing a time when significant increases in beta-cell mass occur, pregnancy was later induced in these CFDA-SE-treated mice to assess the phenotype and fate of the insulin-positive cells in ducts. We found that a small portion of CFDA-SE-labeled duct cells became insulin-positive, but they were not fully functional beta-cells based on the in vitro glucose response and the expression levels of key beta-cell genes. Moreover, these insulin-positive cells in ducts expressed significantly lower levels of genes associated with extracellular matrix degradation and cell migration, which may thus prevent their budding and migration into preexisting islets. A similar conclusion was reached through analysis of the Gene Expression Omnibus database for both mice and humans. Together, our data suggest that the contribution of duct cells to normal beta-cells in adult islets is minimal at best.
Collapse
Affiliation(s)
- Qun Liu
- Department of Endocrinology, The First Affiliated Hospital of NanChang University, Nanchang, 330006, China.,Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15224, USA
| | - Yinan Jiang
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15224, USA
| | - Lingyan Zhu
- Department of Endocrinology, The First Affiliated Hospital of NanChang University, Nanchang, 330006, China.
| | - Jieqi Qian
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15224, USA.,Department of Pediatric Endocrinology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Chaoban Wang
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15224, USA.,Department of Pediatric Endocrinology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Tianlun Yang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, 410078, China
| | - Krishna Prasadan
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15224, USA
| | - George K Gittes
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15224, USA
| | - Xiangwei Xiao
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15224, USA.
| |
Collapse
|
48
|
González-Moro I, Santin I. Long non-coding RNA-regulated pathways in pancreatic β cells: Their role in diabetes. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 359:325-355. [PMID: 33832652 DOI: 10.1016/bs.ircmb.2021.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Long non-coding RNAs (lncRNAs) are transcripts of more than 200 nucleotides that have not coding potential, but act as gene expression regulators through several molecular mechanisms. Several studies have identified tons of lncRNAs that are expressed in pancreatic β cells and many of them have been shown to have β cell-specific expression, suggesting a potential role in the regulation of basal β cell functions. Indeed, accumulating evidence based on numerous studies, has highlighted the implication of lncRNAs in the regulation of pancreatic β cell differentiation and proliferation, insulin synthesis and secretion, and apoptosis. In addition, several lncRNAs have shown to be implicated in pancreatic β cell dysfunction linked to different types of diabetes, including type 1 and type 2 diabetes, and monogenic forms of the disease. Pathogenic conditions linked to diabetes (inflammation or lipoglucotoxicity, for example) dysregulate the expression of several lncRNAs, suggesting that changes in lncRNA may alter potentially important pathways for β cell function, and eventually leading to β cell dysfunction and diabetes development. In this sense, functional characterization of some lncRNAs has demonstrated that these non-coding molecules participate in the regulation of several crucial pathways at the pancreatic β cell level, and dysregulation of these pathways leads to pathogenic phenotypes. In this review, we provide an overview of the action mechanisms of functionally characterized lncRNAs in healthy β cells and describe the contribution of some diabetes-associated lncRNAs to pancreatic β cell failure.
Collapse
Affiliation(s)
- Itziar González-Moro
- Department of Biochemistry and Molecular biology, University of the Basque Country, Leioa, Spain; Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Izortze Santin
- Department of Biochemistry and Molecular biology, University of the Basque Country, Leioa, Spain; Biocruces Bizkaia Health Research Institute, Barakaldo, Spain; CIBER (Centro de Investigación Biomédica en Red) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
49
|
Abstract
Pancreatic islet beta cells (β-cells) synthesize and secrete insulin in response to rising glucose levels and thus are a prime target in both major forms of diabetes. Type 1 diabetes ensues due to autoimmune destruction of β-cells. On the other hand, the prevailing insulin resistance and hyperglycemia in type 2 diabetes (T2D) elicits a compensatory response from β-cells that involves increases in β-cell mass and function. However, the sustained metabolic stress results in β-cell failure, characterized by severe β-cell dysfunction and loss of β-cell mass. Dynamic changes to β-cell mass also occur during pancreatic development that involves extensive growth and morphogenesis. These orchestrated events are triggered by multiple signaling pathways, including those representing the transforming growth factor β (TGF-β) superfamily. TGF-β pathway ligands play important roles during endocrine pancreas development, β-cell proliferation, differentiation, and apoptosis. Furthermore, new findings are suggestive of TGF-β's role in regulation of adult β-cell mass and function. Collectively, these findings support the therapeutic utility of targeting TGF-β in diabetes. Summarizing the role of the various TGF-β pathway ligands in β-cell development, growth and function in normal physiology, and during diabetes pathogenesis is the topic of this mini-review.
Collapse
Affiliation(s)
- Ji-Hyun Lee
- Cell Growth and Metabolism Section, Diabetes, Endocrinology & Obesity Branch, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Clinical Research Center, Bethesda, MD, USA
| | - Ji-Hyeon Lee
- Cell Growth and Metabolism Section, Diabetes, Endocrinology & Obesity Branch, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Clinical Research Center, Bethesda, MD, USA
| | - Sushil G Rane
- Cell Growth and Metabolism Section, Diabetes, Endocrinology & Obesity Branch, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Clinical Research Center, Bethesda, MD, USA
- Correspondence: Sushil G. Rane, PhD, Cell Growth and Metabolism Section, Diabetes, Endocrinology and Obesity Branch, National Institutes of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Clinical Research Center, Building 10, CRC-West 5-5940, 10 Center Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
50
|
Omotehara T, Naito M, Hayashi S, Kawata S, Shimada K, Itoh M. Common hepatic artery originating from superior mesenteric artery with replaced right hepatic artery. Anat Sci Int 2021; 96:568-571. [PMID: 33420976 DOI: 10.1007/s12565-020-00599-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 12/26/2020] [Indexed: 10/22/2022]
Abstract
We found a variational case relating to the arteries distributed to the liver and pancreas during the routine cadaver dissection course. The common hepatic artery originated from the superior mesenteric artery as the first branch. The common hepatic artery was soon divided into the left and right hepatic arteries and distributed to the left and right lobe of the liver. The superior pancreaticoduodenal artery arose from the gastroduodenal artery, a branch of the left hepatic artery. The inferior pancreaticoduodenal artery originated from the superior mesenteric artery. Besides, two posterior branches of the right hepatic artery supplied the duodenum and the head of the pancreas. The arterial arcades were formed behind the head of the pancreas among the superior pancreaticoduodenal artery and these two posterior branches of the right hepatic artery. In conclusion, this case can be considered a rare case in which the replaced common hepatic artery and replaced right hepatic artery occurred simultaneously. The coexisting of these two replaced arteries suggests that the developmental processes to form these variations are independent.
Collapse
Affiliation(s)
- Takuya Omotehara
- Department of Anatomy, Tokyo Medical University, 6-1-1, Shinjuku, Shinjuku-Ku, Tokyo, 160-8402, Japan.
| | - Michiko Naito
- Division of Anatomical Science, Department of Functional Morphology, Nihon University School of Medicine, 30-1, Oyaguchi Kami-cho, Itabashi-Ku, Tokyo, 173-8610, Japan
| | - Shogo Hayashi
- Division of Basic Medical Science, Department of Anatomy, Tokai University School of Medicine, 143, Shimokasuya, Isehara-shi, 259-1193, Kanagawa, Japan
| | - Shinichi Kawata
- Department of Anatomy, Tokyo Medical University, 6-1-1, Shinjuku, Shinjuku-Ku, Tokyo, 160-8402, Japan
| | - Kazuyuki Shimada
- Department of Anatomy, Tokyo Medical University, 6-1-1, Shinjuku, Shinjuku-Ku, Tokyo, 160-8402, Japan.,Division of Anatomical Science, Department of Functional Morphology, Nihon University School of Medicine, 30-1, Oyaguchi Kami-cho, Itabashi-Ku, Tokyo, 173-8610, Japan
| | - Masahiro Itoh
- Department of Anatomy, Tokyo Medical University, 6-1-1, Shinjuku, Shinjuku-Ku, Tokyo, 160-8402, Japan
| |
Collapse
|